4.6 Article

Integration of Metabolomics and Gene Expression Profiling Elucidates IL4I1 as Modulator of Ibrutinib Resistance in ABC-Diffuse Large B Cell Lymphoma

期刊

CANCERS
卷 13, 期 9, 页码 -

出版社

MDPI
DOI: 10.3390/cancers13092146

关键词

DLBCL; lymphoma; transcriptomic; metabolomics; drug resistance; ibrutinib; amino-acid metabolism; metabolic reprogramming

类别

资金

  1. National Institute of General Medical Sciences of the National Institutes of Health [R35GM133510]
  2. Ohio State university

向作者/读者索取更多资源

This study investigates the modulator of ibrutinib resistance in ABC diffuse large B-cell lymphoma by integrating Metabolomics and Gene expression profiling, revealing key genes and pathways driving metabolic deregulation in cancer cells. The findings highlight the role of metabolic reprogramming during drug resistance development, and how drug-resistant clones shift towards oxidative phosphorylation modulated via the BTK-PI3K-AKT-IL4I1 axis.
Simple Summary In this study, we present a workflow to understand the modulator of ibrutinib resistance in ABC diffuse large B cell lymphoma by integrating Metabolomics and Gene expression profiling as shown in the graphical abstract. We performed an untargeted metabolomics analysis using a Q-Exactive high-resolution mass spectrometer to dissect the metabolic reprogramming associated with acquired ibrutinib resistance in paired ibrutinib-sensitive and ibrutinib-resistant DLBCL cell lines. Further, we identified common denominators, integrating metabolome and transcriptome data, confirming clinical significance, integrating pathways, and identifying the candidate gene driving ibrutinib resistance and metabolic reprogramming. Our work demonstrates that a multi-omics approach can be a robust and impartial strategy to uncover genes and pathways that cause metabolic deregulation in cancer cells. Diffuse large B-cell lymphoma (DLBCL) is the most common non-Hodgkin lymphoma (NHL). B-cell NHLs rely on Bruton's tyrosine kinase (BTK) mediated B-cell receptor signaling for survival and disease progression. However, they are often resistant to BTK inhibitors or soon acquire resistance after drug exposure resulting in the drug-tolerant form. The drug-tolerant clones proliferate faster, have increased metabolic activity, and shift to oxidative phosphorylation; however, how this metabolic programming occurs in the drug-resistant tumor is poorly understood. In this study, we explored for the first time the metabolic regulators of ibrutinib-resistant activated B-cell (ABC) DLBCL using a multi-omics analysis that integrated metabolomics (using high-resolution mass spectrometry) and transcriptomic (gene expression analysis). Overlay of the unbiased statistical analyses, genetic perturbation, and pharmaceutical inhibition was further used to identify the key players contributing to the metabolic reprogramming of the drug-resistant clone. Gene-metabolite integration revealed interleukin four induced 1 (IL4I1) at the crosstalk of two significantly altered metabolic pathways involved in producing various amino acids. We showed for the first time that drug-resistant clones undergo metabolic reprogramming towards oxidative phosphorylation and are modulated via the BTK-PI3K-AKT-IL4I1 axis. Our report shows how these cells become dependent on PI3K/AKT signaling for survival after acquiring ibrutinib resistance and shift to sustained oxidative phosphorylation; additionally, we outline the compensatory pathway that might regulate this metabolic reprogramming in the drug-resistant cells. These findings from our unbiased analyses highlight the role of metabolic reprogramming during drug resistance development. Our work demonstrates that a multi-omics approach can be a robust and impartial strategy to uncover genes and pathways that drive metabolic deregulation in cancer cells.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.6
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据